Sunteți pe pagina 1din 16

16

ISSN: 2347-7881
Review Article

Review on Stability Indicating Assay Methods (SIAMs)


Salma S. Quadri*, Lalit V. Sonwane, Bhagwat N. Poul, Sharada N. Kamshette
Department of Quality Assurance,
MSS’s Maharashtra College of Pharmacy, Nilanga,
Latur, Maharashtra, India.
*Salmaq13@gmail.com

ABSTRACT
The main contemporary goal of stability indicating methods is to provide information about condition
for stress testing so as to establish the stability of drug substances and product. This paper reviews the
regulatory aspects for development of stability indicating methods. SIMs are used to differentiate the
API from its potential decomposition product. Regulatory guidance in ICH Q1A (R2) ICH Q3B (R2) Q6A
and FDA 21 CFR section 211 requires validated stability indicating methods. Force degradation is
required to demonstrate the specificity when developing SIMs and for this reason, it should be perform
prior to implementing the stability studies. Force degradation of drug standard and excipients is carried
out under different conditions to determine whether the analytical method is stability indicating. The
approaches for the development of stability indicating method is discussed.

Keywords: Stability indicating method, Regulatory guidelines, Force degradation, Development of


SIAMs, Stress testing

INTRODUCTION which further helps in determination of the


Chemical stability of pharmaceutical molecules intrinsic stability of the molecule, establishing
is a matter of great concern as it affects the degradation pathways and to validate the
safety and efficacy of the drug product. The FDA stability indicating procedures used [1]. But
and ICH guidance states the requirement of these guidelines are very general in conduct of
stability testing data to understand how the forced degradation and do not provide details
quality of a drug substance and drug product about the practical approach towards stress
changes with time under the influence of testing. Although forced degradation studies
various environmental factors. Knowledge of are a regulatory requirement and scientific
the stability of molecule helps in selecting necessity during drug development, it is not
proper formulation and package as well as considered as a requirement for formal stability
providing proper storage conditions and shelf program. It has become mandatory to perform
life, which is essential for regulatory stability studies of new drug moiety before
documentation. Forced degradation is a process filing in registration dossier. The stability studies
that involves degradation of drug products and include long term studies (12 months) and
drug substances at conditions more severe than accelerated stability studies (6 months). But
accelerated conditions and thus generates intermediate studies (6 months) can be
degradation products that can be studied to performed at conditions milder than that used
determine the stability of the molecule. The ICH in accelerated studies. So the study of
guideline states that stress testing is intended degradation products like separation,
to identify the likely degradation products identification and quantitation would take even
How to cite this article: SS Quadri, LV Sonwane, B Poul, S Kamshette; Review on Stability Indicating Assay
Methods (SIAMs); PharmaTutor; 2014; 2(8); 16-31
PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org
17

ISSN: 2347-7881
more time. As compared to stability studies, biotechnological substances and products are
forced degradation studies help in generating also delved upon.
degradants in much shorter span of time, According to FDA guideline (Guidance for
mostly a few weeks. The samples generated Industry, Analytical Procedures and Methods
from forced degradation can be used to Validation, FDA, 2000), a Stability Indicating
develop stability indicating method which can Method (SIM) is defined as a validated
be applied latter for the analysis of samples analytical procedure that accurate and precisely
generated from accelerated and long term measure active ingredients (drug substance or
stability studies. This review provides a proposal drug product) free from process impurities,
on the practical performance of forced excipients and degradation products. The FDA
degradation and its application for the recommends that all assay procedures for
development of stability indicating method. The stability should be stability indicating. The main
stability-indicating assay is a method that is objective of a stability indicating method is to
employed for the analysis of stability samples in monitor results during stability studies in order
pharmaceutical industry. With the advent of to guarantee safety, efficacy and quality. It
International Conference on Harmonisation represents also a powerful tool when
(ICH) guidelines, the requirement of investigating out-of-trend (OOT) [2] or out-of-
establishment of stability-indicating assay specification (OOS) [3] results in quality control
method (SIAM) has become more clearly processes.
mandated. The guidelines explicitly require
conduct of forced decomposition studies under REGULATORY STATUS OF STABILITY-
a variety of conditions, like pH, light, oxidation, INDICATING ASSAYS
dry heat, etc. and separation of drug from The ICH guidelines have been incorporated as
degradation products. The method is expected law in the EU, Japan and in the US, but in
to allow analysis of individual degradation reality, besides these other countries are also
products. A review of literature reveals a large using them. As these guidelines reflect the
number of methods reported over the period of current inspectional tendencies, they carry the
last 3–4 decades under the nomenclature de facto force of regulation. The ICH guideline
‘stability-indicating’. However, most of the Q1A on Stability Testing of New Drug
reported methods fall short in meeting the Substances and Products [4] emphasizes that the
current regulatory requirements. Accordingly, testing of those features which are susceptible
the purpose of this write-up is to suggest a to change during storage and are likely to
systematic approach for the development of influence quality, safety and/or efficacy must be
validated SIAMs that should meet the current done by validated stability-indicating testing
ICH and regulatory requirements. The methods. It is also mentioned that forced
discussion also touches upon various critical decomposition studies (stress testing) at
issues, such as the extent of separation of temperatures in 10 °C increments above the
degradation products, establishment of mass accelerated temperatures, extremes of pH and
balance, etc., which are important with respect under oxidative and photolytic conditions
to the development of stability-indicating should be carried out on the drug substance so
assays, but are not yet fully resolved. Some as to establish the inherent stability
other aspects like suitability of pharmacopoeial characteristics and degradation pathways to
methods for the purpose and the role of SIAMs support the suitability of the proposed
in stability evaluation of biological/ analytical procedures. The ICH guideline Q3B
entitled ‘Impurities in New Drug Products’

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


18

ISSN: 2347-7881
emphasizes on providing documented evidence components of interest can be accurately
that analytical procedures are validated and measured without interference.’ The major
suitable for the detection and Quantitation of changes brought in the new guideline are with
[5]
degradation products . It is also required that respect to (i) introduction of the requirement of
analytical methods should be validated to validation, and (ii) the requirement of analysis
demonstrate that impurities unique to the new of degradation products and other components,
drug substance do not interfere with or are apart from the active ingredients(s). The
separated from specified and unspecified requirement is also listed in World Health
degradation products in the drug product. The Organization (WHO), European Committee for
ICH guideline Q6A, which provides note for Proprietary Medicinal Products and Canadian
guidance on specification [6] also, mentions the Therapeutic Products Directorate’s guidelines
requirement of stability-indicating assays under on stability testing of well established or
Universal Tests/Criteria for both drug existing drug substances and products [10,11,12].
substances and drug products. The same is also Even the United States Pharmacopoeia (USP)
a requirement in the guideline Q5C on Stability has a requirement listed under ‘Stability Studies
Testing of Biotechnological/Biological Products in Manufacturing’, which says that samples of
[7]
. Since there is no single assay or parameter the products should be assayed for potency by
that profiles the stability characteristics of such the use of a stability-indicating assay [13]. The
products, the onus has been put on the requirement in such explicit manner is,
manufacturer to propose a stability-indicating however, absent in other pharmacopoeias.
profile that provides assurance on detection of
changes in identity, purity and potency of the Current ICH guideline on Good Manufacturing
product. Unfortunately, none of the ICH Practices for Active Pharmaceutical Ingredients
guidelines provides an exact definition of a (Q7A), which is under adoption by WHO, also
stability-indicating method. Elaborate clearly mentions that the test procedures used
definitions of stability-indicating methodology in stability testing should be validated and be
are, however, provided in the United States- stability- indicating [14].
Food and Drug Administration (US-FDA) stability
guideline of 1987 [8] and the draft guideline of OBJECTIVE OF STABILITY STUDIES
1998 [9]. Stability-indicating methods according Stability studies are performed to establish the
to 1987 guideline were defined as the shelf life and storage condition of API and
‘quantitative analytical methods that are based product. In recently adopted stability
on the characteristic structural, chemical or guidelines, the committee for proprietary
biological properties of each active ingredient medicinal product (CPMP) indicates the
of a drug product and that will distinguish each objective of stability testing is to provide
active ingredient from its degradation products evidence on how much quality of an API varies
so that the active ingredient content can be with time under influence of the variety of
accurately measured.’ This definition in the environmental factor such as temperature,
draft guideline of 1998 reads as: ‘ validated humidity and light. The stability of API does not
quantitative analytical methods that can detect mean “fix” or “not likely change” but it means
the changes with time in the chemical, physical, “controlled and acceptable change”. Force
or microbiological properties of the drug degradation condition, stress agent
substance and drug product, and that are concentration and time of stress are to be
specific so that the contents of active establishing in such a way that, they effect
ingredient, degradation products, and other degradation preferably 10-20% of parent

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


19

ISSN: 2347-7881
constituent. Stability testing is performed for which stress tests should be carried out – that
welfare of the patient and to protect is, how much stress should be applied or how
reputation of producer, as a requirement of much degradation should be aimed for. In fact,
regulatory agencies to provide data that may be there is not a “gold rule” that attends this issue
of value in formulation of other product [15]. and therefore, it is important to keep in mind
that experimental conditions of stress tests,
STEPS INVOLVED DURING THE DEVELOPMENT should be realistic and lead to “purposeful
OF STABILITY–INDICATING ANALYTICAL degradation” [17].
METHODS (SIAMs) Stress tests should generate representative
A SIAMs is an estimative analytical method used samples to assess drug substance and drug
to detect a trace level amount or residual levels product stability, provide information about
of the API present due to degradation or possible degradation pathway and demonstrate
designing of its synthesis route. As per the FDA the stability indicating power of the analytical
regulations, a SIAMs is defined as a completely procedures applied.
validated method that accurately and precisely
measures API free from potential interferences 1) DETERMINATION OF LIMIT OF
like degradants, biproducts, intermediates, and QUANTIFICATION (LOQ)
excipients and the FDA recommend that all In close relation to the determination of the
assay content methodologies for stability amount of degradation is the evaluation of
[16]
studies be stability indicating . There are Limit of Detection (LOD) and Limit of
three components necessary for implementing Quantification (LOQ) of the method. These
a SIAMs. limits should be closely related to the
1. Generation of degraded samples for testing Reporting, Identification and Qualification of
selectivity of the method, degradation products, as stated in ICH Q3B (R2)
[5]
2. Method development, . These thresholds are determined either as
3. Method validation percentage of drug substance or total daily
intake (TDI) of degradation product. The
Step 1: Generation of degraded samples for analytical methods are usually expected to be
testing selectivity of the method validated for the ability to quantify potential
Here lies one of the main concerns related to a degradation products and drug impurities with
development of a SIM, since the available a LOD and LOQ at least as sensitive as the ICH
guidance documents do not state the extent to threshold (see Figure 1).

Figure 1: ICH thresholds for degradation products in New Drug Application (ICH Q3B)

Reporting threshold
1
Maximum daily dose Threshold2,3
≤1g 0.1%
>1g 0.05%

Identification threshold
Maximum daily dose1 Threshold 2,3
<1mg 1.0% or 5μg TDI, whichever is lower
1mg-10mg 0.5% or 20μg TDI, whichever is lower
>10mg-2g 0.2% or 2mg TDI, whichever is lower 0.10%

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


20

ISSN: 2347-7881

Qualification threshold
1
Maximum daily dose Threshold 2,3
<10 mg 1.0% or 50μg TDI, whichever is lower
10mg – 100 mg 0.5% or 200μg TDI, whichever is lower
>100mg-2g 0.2% or 3mg TDI, whichever is lower
>2g 0.15%

Note: would constitute a type of OOT, the use of a


1. The amount of drug substance administered well studied and well determined LOQ in a SIM,
per day will help the applicator to decide if additional
2. Threshold for degradation product are action are needed to investigate a new
express either as percentage of the drug substance or a OOT.
substance or as total daily intake (TDI) of the
degradation product. lower threshold can be It should be mentioned that these thresholds
appropriate if the degradation product are established for new drug products or New
unusually toxic Drug Application (NDA). For Abbreviated New
3. Higher threshold should be scientifically Drug Application (ANDA) or generic drugs, there
justified. are not specific regulations about this topic and
even less, the companies dealing with these
The identification threshold (IT) varies from 0.1 products, have background information as
to 1.0% of the labeled amount of active those obtained in the development of NDA.
ingredient in the dosage form, or from 5μg to 2 Such application is expected to contain a “full
mg TDI, depending on the maximum daily description of the drug substance including its
dosage in the product´s professional labeling. physical and chemical characteristics and
The identification threshold may be lowered for stability as well; such specifications and
degradation products that may be exceptionally analytical methods are necessary to assure the
toxic. The Reporting Threshold (RT) is either identity, strength, quality, purity and
0.1% or 0.05% depending on the maxim daily bioavailability of drug product and stability data
dosage. For very low dose drug products, where with proposed expiration date”. As already
this type of sensitivity is not attainable, even cited, for ANDA, there are not specific
after exhaustive tentative, justification may be regulations and the same ICH recommendation
provided describing the failed reports. Process- has been used. However, precisely because of
related drug substance impurities that are also the lack of information derived from the new
degradation products should have the same drug development, the complexity and
limits as for ICH Q3B. Ideally, the same responsibility in developing/validating a SIM for
analytical methodology should be used for an ANDA is high. Information like aqueous
Quality Control and Stability Studies. The solubility, pH versus solubility profile, excipients
determination of Out-of-Specification or Out-of- compatibility studies, etc, all information that
Trend results should be more reliable, when enable fully assume the knowledge of the
using a SIM, since LOD and LOQ used allows product ,will help to ensure that best (more
detection of impurities and/or degradation appropriate) condition were chosen for
products adequately. In the situation in which a developing a SIM, like those related to the
new peak arises during stability study and one forced degradation design.
may expect that it should not exist and hence it

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


21

ISSN: 2347-7881
2) Overstressing / Understressing stress, it is recommended to increase
Care should be taken in order to avoid concentrations to 1 Mol L-1 or higher; for
overstressing or understressing samples, with oxidation stress, increase peroxide
may lead to non representative or non- concentration to 10% or 20% (v/v) and/or time
purposeful degradation. So, the use of a of reaction, as well as temperature. If co-
properly designed and executed forced solvents are necessary to increase solubility, it is
degradation study will generate representative recommended the use of acetonitrile that does
samples that will help to ensure that resulting not work as a sensitizer in photostability stress.
method reflects adequately long-term stability Data needs to be evaluated as unusual
[18]
. About the forced degradation (or stress test, degradants may form with co-solvents. If even
both terms will be used in the text) design, it is not all conditions may cause degradation,
[19]
recommended to include alkaline and acidic document efforts and severity of conditions and
hydrolysis, photolysis, oxidation, humidity and should be include in final report. By the other
temperature stress. An compilation of data side, if too much degradation is detected, the
[19-23]
from literature is shown at Table 1 and severity of conditions may be decreased, by
compiles the more often used conditions to diluting acid/bases, neutralizing, reducing
perform forced degradation studies. These exposure time. Also, need to be clarified, that
conditions can be used as a starting point in the synthesis impurities when are not also
development of a SIM. Changing conditions to degradation products do not need to be
harsher or softer levels, can be applied, when described in Stability Studies, but SIM may
too little or too much degradation are obtained. assure that these impurities do not interfere on
For example, in cases in which too little degradation products determination.
degradation was obtained in the hydrolyses

Table 1: “More often” used conditions for forced degradation studies

Solid State

Stress Condition Period of time

Heat 60° C Up to 1 month


Humidity 75% RH Up to 1 month
Photostability 3 mm (powder) Follow ICH requirements
Exposed and non-exposed (Q1B)
samples (“control”)

Solution State

Stress Condition Period of time


Hydrolysis Acid 0.1 – 1 Mol L-1 HCl Up to weeks and 60° C
alkaline 0.1 – 1 Mol L-1 NaOH Up to weeks and 60° C
Oxidation H2O2 3% (v/v) Up to 24 hours
Photostability Exposed and non-exposed Follow ICH requirements
samples (“control”) (Q1B)

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


22

ISSN: 2347-7881
Heat 60° C Up to 1 month

3) FORCED DEGRADATION STUDIES (STRESS conducted on drug substance to obtain


STUDIES) sufficient time for identification of degradation
Forced degradation or stress studies are products and structure elucidation as well as
undertaken to deliberately degrade the sample. optimizing the stress conditions. An early stress
These studies are used to evaluate an analytical study also gives timely recommendations for
method’s ability to measure an active making improvements in the manufacturing
ingredient and its degradation products, process and proper selection of stability-
without interference, by generating potential indicating analytical procedures [23,25].
degradation products. During validation of the
method, drug substance are exposed to acid, LIMITS FOR DEGRADATION
base, heat, light and oxidizing agent to produce The question of how much degradation is
approximately 10% to 30% degradation of sufficient has been the topic of many
active substance. The studies can also provide discussions amongst pharmaceutical scientists.
information about the degradation pathways Degradation of drug substances between 5% to
and degradation products that could form 20% has been accepted as reasonable for
during storage. These studies may also help in validation of chromatographic assays [26,27].
the formulation development, manufacturing, Some pharmaceutical scientists think 10%
and packaging to improve a drug product. degradation is optimal for use in analytical
Reasons for carrying out forced degradation validation for small pharmaceutical molecules
studies include: development and validation of for which acceptable stability limits of 90% of
stability-indicating methodology, determination label claim is common [28]. Others suggested
of degradation pathways of drug substances that drug substance spiked with a mixture of
and drug products, discernment of degradation known degradation products can be used to
products in formulations that are related to challenge the methods employed for
drug substances versus those that are related to monitoring stability of drug product [22]. No such
non–drug substances (e.g., excipients) [17,22]. limits for physiochemical changes, loss of
activity or degradation during shelf life have
APPROPRIATE TIMING been established for individual types or groups
It is very important to know when to perform of biological products [29].
forced degradation studies for the development
of new drug substance and new drug product. It is not necessary that forced degradation
FDA guidance states that stress testing should would result in a degradation product. The
be performed in Phase III of regulatory study can be terminated if no degradation is
submission process. Stress studies should be seen after drug substance or drug product has
done in different pH solutions, in the presence been exposed to stress conditions than that
of oxygen and light, and at elevated conditions mentioned in accelerated stability
temperatures and humidity levels to determine protocol [30]. This is indicative of the stability of
the stability of the drug substance. These stress the molecule under test. Over-stressing a
studies are conducted on single batch. The sample may lead to the formation of secondary
results should be summarized and submitted in degradation product that would not be seen in
an annual report [24]. However, starting stress formal shelf life stability studies and under-
testing early in preclinical phase or phase I of stressing may not generate sufficient
clinical trials is highly encouraged and should be degradation products [31]. Protocols for

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


23

ISSN: 2347-7881
generation of product-related degradation may
differ for drug substance and drug product due Forced degradation is only considered complete
to differences in matrices and concentrations. It after the manufacturing process is finalized,
is recommended that maximum of 14 days for formulations established, and test procedures
stress testing in solution (a maximum of 24 h for developed and qualified. The conditions listed
oxidative tests) to provide stressed samples for in Table 1 are by no means exhaustive and
methods development [32]. should be adjusted by the researcher as needed
to generate ~10% degradation of the API.
EXPERIMENTAL DESIGN
In designing forced degradation studies, it must The nature (inherent stability/instability) of the
be remembered that more strenuous particular drug substance will determine in
conditions than those used for accelerated which direction to adjust the stress conditions.
studies (25°C/60% RH or 40°C/75% RH) should Also, the aforementioned conditions could be
be used. At a minimum, the following used to stress the drug substance or drug
conditions should be investigated: product either in the solid or liquid/suspension
(1) Acid and base hydrolysis, form as applicable.
(2) Hydrolysis at various ph,
(3) Thermal degradation, For oxidative degradation with H2O2, at least
(4) Photolysis, and one of the storage conditions should be at room
(5) Oxidation. For the drug substance and drug temperature. Heating H2O2 solution increases
product, the scheme shown in Figure 2 could be the homolytic cleavage of the HO-OH bond to
used as a guide. form the alkoxy radical. The alkoxy radical is
very reactive and may come to dominate the
The initial experiments should be focused on observed degradation pathway. Adding a small
determining the conditions that degrade the quantity of methanol in a confirmatory stress
drug by approximately 10%. The conditions experiment quenches the alkoxy radical and
generally employed for forced degradation are rules out species produced by this more
summarized in Table 2. aggressive oxidizing agent. Also, the formation
of peroxycarboxymidic acid has been observed
However, some scientists have found it practical when acetonitrile is used as a cosolvent in H2O2
to begin at extreme conditions (80°C or even stress studies (in basic conditions). The
higher, 0.5N NaOH, 0.5N HCl, 3% H2O2) and peroxycarboximidic acid has activated
testing at shorter (2, 5, 8, and 24 hrs, etc) hydroxylation reactivity, which is not
multiple time points, thus allowing for a rough representative of H2O2. To circumvent these
evaluation of rates of degradation. Testing at problems, some research scientists always
early time points may permit distinction perform a parallel or alternative oxidative study
between primary degradants and their using azobisisobutyronitrile (AIBN), which is a
secondary degradation products. This strategy less reactive oxidant and has been shown to
allows for better degradation pathway produce more representative degradants.
determination. It must be noted that a forced
degradation study is a “living process” and List of some common conditions used in
should be done along the developmental time conducting forced degradation studies for drug
line as long as changes in the stability-indicating substances as shown in Table 3.
methods, manufacturing processes, or
formulation changes are ongoing.

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


24

ISSN: 2347-7881

Figure 2: An Illustrative Diagram Showing the Different Forced Degradation Condition to be used for
Drug Substance and Drug Product

Table 2: Conditions Generally Employed For Forced Degradation [24]


Degradation Type Experimental Condition Storage Condition Sampling Time
Control API (no acid or base) 40 0C, 600C 1,3,5 days
0.1N NAOH 40 0C, 600C 1,3,5 days
Hydrolysis Acid Control(no API) 40 0C, 600C 1,3,5 days
Base Control(no API) 40 0C, 600C 1,3,5 days
pH: 2,4,6,8 40 0C, 600C 1,3,5 days
3% H2O2 25 0C, 600C 1,3,5 days
Peroxide control 25 0C, 600C 1,3,5 days
Oxidation Azobisisobutyronitrile (AIBN) 40 0C, 600C 1,3,5 days
AIBN Control 40 0C, 600C 1,3,5 days
Light, 1X ICH NA 1,3,5 days
Photolytic Light, 3X ICH NA 1,3,5 days
Light Control NA 1,3,5 days
Heat chamber 60 0C 1,3,5 days
Thermal Heat chamber 60 0C /75% RH 1,3,5 days
Heat chamber 80 0C 1,3,5 days
Heat chamber 80 0C /75% RH 1,3,5 days
Heat control Room Temp. 1,3,5 days

Table 3: lists some common conditions used in conducting forced degradation studies for drug
substances

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


25

ISSN: 2347-7881
Sample condition Time / Exposure
Solid / 60 - 70ºC 7 – 10 days
Solid / 60 - 70ºC / 75% RH 10 days
Solid / simulated sunlight 2 – 3 weeks x ICH
confirmatory exposure
0.1 to 2 N HCl solutions either at RT or at 60 - 70ºC 1 – 3 days
0.1 to 1 N NaOH solutions either at RT or at 60 - 70ºC 1 – 3 days
Dilute hydrogen peroxide (0.1 to 6%) at RT or at 60 - 70ºC 1 – 3 days
Solution in Water or at 60 - 70ºC 1 – 3 days

SELECTION OF DRUG CONCENTRATION at a concentration which the drug is expected


Which concentration of drug should be used for to be present in the final formulations [34]. The
degradation study has not been specified in reason for proposing this are the examples of
regulatory guidance. It is recommended that aminopenicillins and aminocephalosporins
the studies should be initiated at a where a range of polymeric products have been
concentration of 1 mg/ml [33]. By using drug found to be formed in commercial preparations
concentration of 1 mg/ml, it is usually possible of containing drug in high concentrations [35].
to get even minor decomposition products in Example-The goal is to degrade the active
the range of detection. It is suggested that moiety to 5-10% in sample for which the
some degradation studies should also be done conditions can be used as Shown in Table 4.

Table 4: Conditions for different pH


Study Conditions
Acidic pH 0.1N HCl
Neutral pH pH 7.0 phosphate
buffer
Basic pH 0.1N NaOH
Oxidation O2 Atmosphere, H2O2
Photolysis (UV) 1000 Watts Hrs/M2
Photolysis 6 x 106 Lux Hrs
(Fluorescence)

METHOD DEVELOPMENT AND OPTIMIZATION various ratios for the initial stages of separation.
Before starting method development, various Selection between methanol and acetonitrile
physiochemical properties like pKa value, log P, for organic phase is based on the solubility of
solubility and absorption maximum of the drug the analyte. Initially the water: organic phase
must be known for it lays a foundation for HPLC ratio can be kept at 50: 50 and suitable
method development. Log P and solubility helps modifications can be made as trials proceed to
select mobile phase and sample solvent while obtain a good separation of peaks. Latter buffer
pKa value helps determine the pH of the mobile can be added if it is required to obtain better
phase [34]. Reverse phase column is a preferred peak separation and peak symmetry. If the
choice to start the separation of sample method is to be extended to LC-MS then mobile
components as the degradation is carried out in phase buffer should be MS compatible like
aqueous solution. Methanol, water and triflouroacetic acid and ammonium formate.
acetonitrile can be used as mobile phase in Variation in column temperature affects the

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


26

ISSN: 2347-7881
selectivity of the method as analytes respond acceptance criteria for validation, the method is
differently to temperature changes. A modified and revalidated [36].
temperature in the range of 30-40°C is suitable Validation is defined by the International
[16]
to obtain good reproducibility . It is better to Organization for Standardization (ISO) as
push the drug peak farther in chromatogram as “verification, where the specified requirements
it results in separation of all degradation are adequate for an intended use”, where the
products. Also a sufficient run time after the term verification is defined as “provision of
drug peak is to be allowed to obtain the objective evidence that a given item fulfills
[33]
degradants peak eluting after the drug peak . specified requirements” [39]. The applicability
During method development it may happen and scope of an analytical method should be
that the drug peak may hide an impurity or defined before starting the validation process. It
degradant peak that co-elutes with the drug. includes defining the analytes, concentration
This requires peak purity analysis which range, description of equipment and
determines the specificity of the method. Direct procedures, validation level and criteria
analysis can be done on line by using photo required. The validated range is defined by
diode array (PDA) detection. PDA provides IUPAC as “the interval of analyte concentration
information of the homogeneity of the spectral within which the method can be regarded as
peak but it is not applicable for the degradants validated” [40,41]. This range does not have to be
that have the similar UV spectrum to the drug. the highest and lowest possible levels of the
Indirect method involves change in the analyte that can be determined by the method.
chromatographic conditions like mobile phase Instead, it is defined on the basis of the
ratio, column, etc. which will affect the peak intended purpose of the method [42,43]. The
separation. The spectrum of altered method can be validated for use as a screening
chromatographic condition is then compared (qualitative), semi-quantitative (e.g. 5-10ppm)
with the original spectra. If the degradant peaks or quantitative method. It can also be validated
and area percentage of the drug peak remains for use on single equipment, different
same then it can be confirmed that the drug equipments in the laboratory, different
peak is homogeneous [36]. The degradant that laboratories or even for international use at
co-elutes with the drug would be acceptable if it different climatic and environmental conditions.
is not found to be formed in accelerated and The criteria of each type of validation will of
[1]
long term storage conditions . The method is course be different with the validation level
then optimized for separating closely eluting required [39,44].
peaks by changing flow rate, injection volume,
column type and mobile phase ratio. APPROACHES TO VALIDATION EXPERIMENTS
Accuracy is established by quantitation of the
METHOD VALIDATION sample against a reference standard for API, or
The developed SIM is then validated according spiking placebo with API for drug product. It can
to USP/ICH guideline for linearity, accuracy, also be determined by comparison of results
precision, specificity, quantitation limit, from alternate measurement techniques.
detection limit, ruggedness and robustness of Precision is determined by multiple
the method. It is required to isolate, identify measurements on an authentic, homogeneous
and quantitate the degradants found to be set of samples. Samples may be analyzed on
above identification threshold (usually 0.1%) different days, by different analysts, on
[37,38]
. If the method does not fall within the different instruments, or in different
laboratories. There are three levels of precision

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


27

ISSN: 2347-7881
validation evaluations – repeatability, tool for examining proportionality of the
intermediate precision, and reproducibility. response. The range is established by the
Repeatability is a measure of precision under required limits of the method and the point at
the same conditions over a short period of time. which linearity is compromised.
Intermediate precision is a measure of precision Robustness is typically assessed by the effect
within the same laboratory by different of small deliberate changes to chromatographic
operators, using different instruments, and methods on system suitability parameters such
making measurements on different days. as peak retention, resolution, and efficiency.
Reproducibility assesses precision between two Experimental factors that are typically varied
or more laboratories. during method robustness evaluations include
[45]
Specificity can be established by a number of :
approaches, depending on the intended (i) Age of standards and sample preparations,
purpose of the method. The ability of the (ii) Sample extraction time,
method to assess the analyte of interest in a (iii) Variations to pH of mobile phase,
drug product is determined by a check for (iv) Variation in mobile phase composition,
interference by placebo. Specificity can be (v) Analysis temperature,
assessed by measurement of the API in samples (vi) Flow rate,
that are spiked with impurities or degradants, if (vii) Column lot and/or manufacturer,
available. If API-related compounds are not (viii) Type and use of filter against
available, drug can be stressed or force- centrifugation. Robustness experiments are an
degraded in order to produce degradation ideal opportunity to utilize statistical design of
products. In chromatographic separations, experiments, providing data driven method
apparent separation of degradants may be control.
confirmed by peak purity determinations by
photodiode array, mass purity determinations The ICH guidance on validation separates types
by mass spectroscopy (MS), or by confirming of methods according to the purpose of the
separation efficiency using alternate column method and lists which evaluations are
chemistry. During forced degradation appropriate for each type [28].
experiments, degradation is targeted at 5 to The ICH guidance also suggests detailed
20% degradation of the API, in order to avoid validation schemes relative to the intended
concerns about secondary degradation. purpose of the methods. It lists recommended
The limit of Detection and limit of Quantitation data to report for each validation parameter.
are based on measurement Signal-to-noise Acceptance criteria for validation elements
ratios of 3 and 10, respectively. Standards or must be based on the historical performance of
samples at concentrations near the expected the method, the product specifications, and
limits are measured. Signal- to-noise can be must be appropriate for the phase of drug
generated by software, manually measured, development.
estimated from standard deviation calculations,
or limits may be empirically determined. OTHER ANALYTICAL METHODS FOR
Linearity is established by measuring response DEVELOPING SIM
at various concentrations by a regression plot, Stability-indicating methods will be
typically by method of least squares. The characterized by potency, purity and biological
response may require mathematical activity [46]. The selection of tests is product
manipulation prior to linearity assessments. A specific. Stability indicating methods may
visual inspection of the linearity plot is the best include various methods like electrophoresis

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


28

ISSN: 2347-7881
(SDS-PAGE, immunoelectrophoresis, Western techniques like Reversed Phase High
blot, isoelectrofoccusing), high-resolution Performance Liquid Chromatography (RP-HPLC),
chromatography (e.g., reversed phase Thin Layer Chromatography (TLC), Gas
chromatography, SEC, gel filtration, ion Chromatography (GC), Capillary Electrophoresis
exchange, and affinity chromatography) and (CE), Capillary Electrophoresis Chromatography
[47]
peptide mapping . The analytical method of (CEC) and Super critical Fluid Chromatography
choice should be sensitive enough to detect (SFC). An excellent combination of hyphenated
impurities at low levels (i.e., 0.05% of the chromatographic and spectroscopic technique
analyte of interest or lower) and the peak such as HPLC-DAD (High Performance Liquid
responses should fall within the range of Chromatography Photodiode Array ultraviolet
detector's linearity. The analytical method Detector), LC-MS (Liquid Chromatography-Mass
should be capable of capturing all the impurities Spectrometry), LC-NMR (Liquid
formed during a formal stability study at or Chromatography-Nuclear Magnetic Resonance)
below ICH threshold limits [48,49]. Degradation and GCMS (Gas Chromatography-Mass
product identification and characterization are Spectrometry) are used when degradants
to be performed based on formal stability cannot be isolated in pure form. HPLC-DAD and
results in accordance with ICH requirements. LC-MS are used to compare the RRT (relative
Conventional methods (e.g., column retention time), UV spectra, mass spectra
chromatography) or hyphenated techniques (MS/MS or MSN) [52]. Ranjit Singh et al.
(e.g., LC–MS, LC–NMR) can be used in the discussed the role of hyphenated systems for
identification and characterization of the the isolation of degradants and impurities [50].
degradation products. Use of these techniques
can provide better insight into the structure of CONCLUSION
the impurities that could add to the knowledge Stability-indicating method is an analytical
space of potential structural alerts for procedure that is capable of discriminating
genotoxicity and the control of such impurities between the major active (intact)
with tighter limits [37,47,50]. It should be noted pharmaceutical ingredients (API) from any
that structural characterization of degradation degradation (decomposition) product(s) formed
products is necessary for those impurities under defined storage conditions during the
formed during formal shelf-life stability studies stability evaluation period.
[48]
and above the qualification threshold limit . Forced degradation studies are indispensable in
the development of stability-indicating and
New analytical technologies that are degradant-monitoring methods as part of a
continuously being developed can also be used validation protocol. Forced degradation studies
when it is appropriate to develop stability also provide invaluable insight in investigating
indicating method [51]. The unknown impurity, degradation products the use of properly
which is observed during the analysis, designed and executed forced degradation
pharmaceutical development, stress studies study will generate a representative sample
and formal stability studies of the drug that will in turn help to develop stability-
substances and drug product, can be separated indicating method.
and analyzed by using various chromatographic

↓ REFERENCES
1. International Conference on Harmonization (ICH) (2003) Harmonised Tripartite Guideline on, Topic
Q1A, Notes for Guidance on stability testing: Stability testing of new drug substances and new drug

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


29

ISSN: 2347-7881
products products. Pub by The European Agency for the Evaluation of Medicinal Products, Human
medicine evaluation Unit.
2. Swartz M, Krull I (2004) Investigating Out-Of-Specifications Results. LCGC 22: 132-136.
3. Center for Drug Evaluation and Research (CDER) (2006) Guidance for Industry Investigating Out-of-
Specification (OOS) Test Results for Pharmaceutical Production.
4. International Conference on Harmonization (ICH) (1993) Harmonised Tripartite Guideline Notes for
Stability Testing of New Drug Substances and Products. Pub by The European Agency for the Evaluation
of Medicinal Products, Human medicine evaluation Unit.
5. International Conference on Harmonization (ICH) (1996) Harmonised Tripartite Guideline Notes for
Impurities in New Drug Products. Pub by The European Agency for the Evaluation of Medicinal Products,
Human medicine evaluation Unit.
6. International Conference on Harmonization (ICH) (1999) Harmonised Tripartite Guideline Notes for
Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and New Drug
Products: Chemical Substances. Pub by The European Agency for the Evaluation of Medicinal Products,
Human medicine evaluation Unit.
7. International Conference on Harmonization (ICH) (1995) Harmonised Tripartite Guideline Notes for
Quality of Biotechnological Products: Stability Testing of Biotechnological/Biological Products. Pub by
The European Agency for the Evaluation of Medicinal Products, Human medicine evaluation Unit.
8. FDA (1987) Guidance for Industry: Submitting Documentation for the Stability of Human Drugs and
Biologics. Food and Drug Administration, Rockville, MD.
9. FDA (1998) Guidance for Industry: Stability Testing of Drug Substances and Drug Products (Draft
guidance), Food and Drug Administration, Rockville, MD.
10. WHO (1996) Guidance for Stability Testing of Pharmaceutical Products Containing Well Established
Drug Substances in Conventional Dosage Forms, in WHO Expert Committee on Specifications for
Pharmaceutical Preparations. Technical Report Series 863, World Health Organization, Geneva, 65–79.
11. CPMP (1998) Guidance on Stability Testing of Existing Active Substances and Related Finished
Products. Committee for Proprietary Medicinal Products, EMEA, London.
12. TPD (1997) Stability Testing of Existing Drug Substances and Products. Therapeutic Products
Directorate, Ottawa.
13. United States Pharmacopeia 24(2000) Rockville, MD: USP Convention Inc.
14. International Conference on Harmonization (ICH) (2000) Harmonised Tripartite Guideline Notes for
Good Manufacturing Practices for Active Pharmaceutical Ingredients. Pub by The European Agency for
the Evaluation of Medicinal Products, Human medicine evaluation Unit.
15. Sehrawat R, Maithani M, Singh R, Regulatory aspects in development of stability indicating method:
A Review. Chromatographia 2010; 72: 1-6.
16. Snyder L.R., Glajch J.L., Kirkland J.J., Practical HPLC method Development. New York: John Wiley
1988; 227-251
17. Ngwa G, Forced Degradation Studies. Forced Degradation as an Integral part of HPLC Stability
Indicating Method Development Drug Delivery Technology 2010.
18. International Conference on Harmonization (ICH) (2003) Harmonised Tripartite Guideline on, Topic
Q1A (R2), Notes for Guidance on Stability Testing of new Drug Substances and Product. Pub by The
European Agency for the Evaluation of Medicinal Products, Human medicine evaluation Unit.
19. Klick S, Muijselaar P, Waterval J, Eichinger T, Korn C, Toward a Generic Approach for Stress Testing of
Drug Substances and Drug Products. Pharm. Technol 2005; 29: 48-57.
20. Alsante K.M., Ando A, Brown R, Ensing J, Hatajik T.D., The Role of Degradant Profiling in Active

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


30

ISSN: 2347-7881
Pharmaceutical Ingredients and Drug Products. Adv. Drug Delivery Rev 2007; 59:29—37.
21. Reynolds D, Forced Degradation of Pharmaceuticals. Am. Pharm 2004; 56-61.
22. Reynolds D, Facchine K, Mullaney F, Alsante K, Hatajik T, Available Guidance and Best Practices for
Conducting Forced Degradation Studies. Pharmaceutical Technology 2002; 48-56.
23. Kats M, International Forced Degradation Studies: Regulatory Considerations and Implementation
2005; 18:1-7.
24. FDA (2003) Guidance for Industry: INDs for Phase II and III Studies – Chemistry, Manufacturing, and
Controls Information, Food and Drug Administration. Rockville, MD.
25. FDA (1999) Guidance for Industry: INDs for Phase 2 and 3 Studies of Drugs, Including Specified
Therapeutic Biotechnology-Derived Products, Draft Guidance, Food and Drug Administration. Rockville,
MD.
26. Szepesi G, Selection of high-performance liquid chromatographic methods in pharmaceutical
analysis. J. Chromatogr 1989; 464: 265-278.
27. Carr G.P., Wahlich J.C., A practical approach to method validation in pharmaceutical analysis. J.
Pharm. Biomed. Anal 1990; 86: 613-618.
28. Jenke D.R., Chromatographic method validation: A review of common practices and procedures II. J.
Liq. Chromatogr 1996; 19: 737-757.
29. International Conference on Harmonization (ICH) (1996) Harmonised Tripartite Guideline on Stability
Testing of Biotechnological/Biological Products Availability. Pub by The European Agency for the
Evaluation of Medicinal Products, Human medicine evaluation Unit.
30. International Conference on Harmonization (ICH) (1996) Harmonised Tripartite Guideline on, Topic
Q1B, Notes for Guidance on Photo stability Testing of New Drug Substances and Product. Pub by The
European Agency for the Evaluation of Medicinal Products, Human medicine evaluation Unit.
31. Maheswaran R, FDA perspectives: scientific considerations of forced degradation studies in ANDA
submissions. Pharm. Technol 2012; 36: 73-80.
32. Klick S, Pim G.M., Waterval J, Thomas E, Alexander J, Toward a generic approach for stress testing of
drug substances and drug products. Pharm. Technol 2005; 29:48–66.
33. Bakshi M, Singh S, Development of validated stability-indicating assay methods-critical review. J.
Pharm. Biomed. Anal 2002; 28: 1011-1040.
34. Singh S, Bakshi M, Guidance on conduct of stress tests to determine inherent stability of drugs.
Pharm. Technol 2000; 24:1-14.
35. Larsen C, Bundgaard H, Polymerization of Penicillins.V. Separation, identification and quantitative
determination of Antigenic Polymerization products in Ampicillin Sodium preparations by high-
performance liquid chromatography. J. Chromatogr 1978; 147: 143–150.
36. Riddhiben M.P., Piyushbhai M.P., Natubhai M.P., Stability indicating HPLC method development – a
review. Int. Res. J. Pharm 2011; 2:79-87.
37. International Conference on Harmonization (ICH) (2000) Harmonised Tripartite Guideline Notes for
Specifications: Test Procedures and Acceptance Criteria for New Drug Substances and New Drug
Products: Chemical Substances. Pub by The European Agency for the Evaluation of Medicinal Products,
Human medicine evaluation Unit.
38. Maha M.A.R., Alia N.W., Sayed S.A., Sayed H.Z., Mohamed A, Validated stability indicating methods
for determination of Nitazoxanide in presence of its degradation products. J. Pharm. Anal 2012; 2:105-
116.
39. ISO/IEC (2007) (1stedn) Guidance on International vocabulary of metrology – Basic and general
concepts and associated terms (VIM). International Organization for Standardization. Geneva.

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org


31

ISSN: 2347-7881
40. Perlatti B, Maria F.D., Gracas F.D.S., Fernandes J.B., Ross M, Validation and application of HPLC–ESI-
MS/MS method for the quantification of RBBR decolorization, a model for highly toxic molecules, using
several fungi strains. Bio Tech 2012; 124:37–44.
41. Thompson M, Ellison S.L.R., Wood R, Harmonized guidelines for single-laboratory validation of
methods of analysis – IUPAC technical report. Pure. Appl. Chem 2002; 74:835-855.
42. Susan D, Van A, Henry J.N., Chinfong C, HPLC method validation studies on a specific assay for
monomethoxypoly (ethylene glycol) succinimido carbonate (mPEG-SC). J Pharmaceut Biomed 2009;
50:138–143.
43. Potts A.R., Tatiana P, Cassandra J, Luba P, Ahalya W, Validation of a quantitative HPLC method for
bacitracin and bacitracin zinc using EDTA as a mobile-phase modifier. J Pharmaceut Biomed Anal 2012;
70:619–623.
44. Hill A.R.C., Reynolds S.L., Guidelines for in-house validation of analytical methods for pesticide
residues in food and animal feeds. Analyst 1999; 124:953-958.
45. International Conference on Harmonization (ICH) (2005) Harmonised Tripartite Guideline on, Topic
Q2 (R1), Notes for Guidance on Validation of Analytical Procedures, Text and Methodology. Pub by The
European Agency for the Evaluation of Medicinal Products, Human medicine evaluation Unit.
46. Bichsel V.E., Curcio V, Gassmann R, Otto H, Requirements for the quality control of chemically
synthesized peptides and biotechnologically produced proteins. Pharm. Acta Helv 1996; 71: 439-446.
47. EMA (2007) Guideline on the Limits of Genotoxic Impurities, Committee for Medical Products for
Human Use (CHMP).
48. International Conference on Harmonization (ICH) (2006) Harmonised Tripartite Guideline on, Topic
Q3A (R2), Notes for Guidance on Impurities in New Drug Substances. Pub by The European Agency for
the Evaluation of Medicinal Products, Human medicine evaluation Unit.
49. International Conference on Harmonization (ICH) (2006) Harmonised Tripartite Guideline on, Topic
Q3B (R2), Notes for Guidance on Impurities in New Drug Products. Pub by The European Agency for the
Evaluation of Medicinal Products, Human medicine evaluation Unit.
50. Singh R, Rehman Z, Current trends in forced degradation study for pharmaceutical product
development. J. Pharm. Educ. Res 2012; 3: 54-63.
51. International Conference on Harmonization (ICH) (1999) Harmonised Tripartite Guideline on, Topic
Q6B, Notes for Guidance on Specifications: Test Procedures and Acceptance Criteria for Bio-
technological/Biological Products. Pub by The European Agency for the Evaluation of Medicinal
Products, Human medicine evaluation Unit.
52. Qiu F, Norwood D.L., Identification of pharmaceutical impurities. J. Liq. Chromatogr 2007; 30:877-
935.

PharmaTutor Magazine | Vol. 2, Issue 8 | magazine.pharmatutor.org

S-ar putea să vă placă și